Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Stem Cells ; 34(3): 523-36, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26840228

RESUMO

In facing the daunting challenge of using human embryonic and induced pluripotent stem cells to study complex neural circuit disorders such as schizophrenia, mood and anxiety disorders, and autism spectrum disorders, a 2012 National Institute of Mental Health workshop produced a set of recommendations to advance basic research and engage industry in cell-based studies of neuropsychiatric disorders. This review describes progress in meeting these recommendations, including the development of novel tools, strides in recapitulating relevant cell and tissue types, insights into the genetic basis of these disorders that permit integration of risk-associated gene regulatory networks with cell/circuit phenotypes, and promising findings of patient-control differences using cell-based assays. However, numerous challenges are still being addressed, requiring further technological development, approaches to resolve disease heterogeneity, and collaborative structures for investigators of different disciplines. Additionally, since data obtained so far is on small sample sizes, replication in larger sample sets is needed. A number of individual success stories point to a path forward in developing assays to translate discovery science to therapeutics development.


Assuntos
Transtorno do Espectro Autista/terapia , Células-Tronco Embrionárias Humanas/transplante , Células-Tronco Pluripotentes Induzidas/transplante , Esquizofrenia/terapia , Transtorno do Espectro Autista/genética , Redes Reguladoras de Genes , Humanos , Transtornos do Humor/genética , Transtornos do Humor/terapia , Esquizofrenia/genética , Pesquisa com Células-Tronco
2.
J Gen Virol ; 93(Pt 11): 2436-2446, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22875256

RESUMO

Congenital human cytomegalovirus (HCMV) infection can cause severe brain abnormalities. Apoptotic HCMV-infected brain cells have been detected in a congenitally infected infant. In biologically relevant human neural precursor cells (hNPCs), cultured in physiological oxygen tensions, HCMV infection (m.o.i. of 1 or 3) induced cell death within 3 days post-infection (p.i.) and increased thereafter. Surprisingly, its known anti-apoptotic genes, including the potent UL37 exon 1 protein (pUL37x1) or viral mitochondria-localized inhibitor of apoptosis (vMIA), which protects infected human fibroblasts (HFFs) from apoptosis and from caspase-independent, mitochondrial serine protease-mediated cell death, were expressed by 2 days p.i. Consistent with this finding, an HCMV UL37x1 mutant, BADsubstitutionUL37x1 (BADsubUL37x1) induced cell death in hNPCs (m.o.i. = 1) to level which were indistinguishable from parental virus (BADwild-type)-infected hNPCs. Surprisingly, although BADsubUL37x1 is growth defective in permissive HFFs, it produced infectious progeny in hNPCs with similar kinetics and to levels comparable to BADwild-type-infected hNPCs (m.o.i. = 1). While delayed at a lower multiplicity (m.o.i. = 0.3), the BADsubUL37x1 mutant reached similar levels to revertant within 12 days, in contrast to its phenotype in HFFs. The inability of pUL37x1/vMIA to protect hNPCs from HCMV-induced cell death did not result from impaired trafficking as pUL37x1/vMIA trafficked efficiently to mitochondria in transfected hNPCs and in HCMV-infected hNPCs. These results establish that pUL37x1/vMIA, although protective in permissive HFFs, does not protect HCMV-infected hNPCs from cell death under physiologically relevant oxygen tensions. They further suggest that pUL37x1/vMIA is not essential for HCMV growth in hNPCs and has different cell type-specific roles.


Assuntos
Citomegalovirus/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Morte Celular , Linhagem Celular , Efeito Citopatogênico Viral , Éxons , Citometria de Fluxo , Regulação Viral da Expressão Gênica , Humanos , Mutação , Oxigênio , Transporte Proteico , RNA Viral/genética , RNA Viral/metabolismo
3.
Stem Cells ; 28(11): 1918-29, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20827750

RESUMO

Medulloblastoma (MDB) is the most common brain malignancy of childhood. It is currently thought that MDB arises from aberrantly functioning stem cells in the cerebellum that fail to maintain proper control of self-renewal. Additionally, it has been reported that MDB cells display higher endogenous Notch signaling activation, known to promote the survival and proliferation of neoplastic neural stem cells and to inhibit their differentiation. Although interaction between hypoxia-inducible factor-1α (HIF-1α) and Notch signaling is required to maintain normal neural precursors in an undifferentiated state, an interaction has not been identified in MDB. Here, we investigate whether hypoxia, through HIF-1α stabilization, modulates Notch1 signaling in primary MDB-derived cells. Our results indicate that MDB-derived precursor cells require hypoxic conditions for in vitro expansion, whereas acute exposure to 20% oxygen induces tumor cell differentiation and death through inhibition of Notch signaling. Importantly, stimulating Notch1 activation with its ligand Dll4 under hypoxic conditions leads to expansion of MDB-derived CD133(+) and nestin(+) precursors, suggesting a regulatory effect on stem cells. In contrast, MDB cells undergo neuronal differentiation when treated with γ-secretase inhibitor, which prevents Notch activation. These results suggest that hypoxia, by maintaining Notch1 in its active form, preserves MDB stem cell viability and expansion.


Assuntos
Neoplasias Encefálicas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Meduloblastoma/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Antígeno AC133 , Antígenos CD/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias Encefálicas/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Glicoproteínas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/metabolismo , Meduloblastoma/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Peptídeos/metabolismo , Reação em Cadeia da Polimerase , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição HES-1 , Células Tumorais Cultivadas
4.
Stem Cells ; 27(1): 7-17, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18832593

RESUMO

Hypoxia commonly occurs in solid tumors of the central nervous system (CNS) and often interferes with therapies designed to stop their growth. We found that pediatric high-grade glioma (HGG)-derived precursors showed greater expansion under lower oxygen tension, typical of solid tumors, than normal CNS precursors. Hypoxia inhibited p53 activation and subsequent astroglial differentiation of HGG precursors. Surprisingly, although HGG precursors generated endogenous bone morphogenetic protein (BMP) signaling that promoted mitotic arrest under high oxygen tension, this signaling was actively repressed by hypoxia. An acute increase in oxygen tension led to Smad activation within 30 minutes, even in the absence of exogenous BMP treatment. Treatment with BMPs further promoted astroglial differentiation or death of HGG precursors under high oxygen tension, but this effect was inhibited under hypoxic conditions. Silencing of hypoxia-inducible factor 1alpha (HIF1alpha) led to Smad activation even under hypoxic conditions, indicating that HIF1alpha is required for BMP repression. Conversely, BMP activation at high oxygen tension led to reciprocal degradation of HIF1alpha; this BMP-induced degradation was inhibited in low oxygen. These results show a novel, mutually antagonistic interaction of hypoxia-response and neural differentiation signals in HGG proliferation, and suggest differences between normal and HGG precursors that may be exploited for pediatric brain cancer therapy.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Glioma/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Criança , Regulação para Baixo/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Mitose/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Oxigênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Proteína Supressora de Tumor p53/metabolismo
5.
J Cell Physiol ; 220(3): 562-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19441077

RESUMO

Oxygen (O2) is a substrate for energy production in the cell and is a rapid regulator of cellular metabolism. Recent studies have also implicated O2 and its signal transduction pathways in controlling cell proliferation, fate, and morphogenesis during the development of many tissues, including the nervous system. O2 tensions in the intact brain are much lower than in room air, and there is evidence that dynamic control of O2 availability may be a component of the in vivo neural stem cell (NSC) niche. At lower O2 tensions, hypoxia-inducible factor 1alpha (HIF1alpha) facilitates signal transduction pathways that promote self-renewal (e.g., Notch) and inhibits pathways that promote NSC differentiation or apoptosis (e.g., bone morphogenetic proteins). Increasing O2 tension degrades HIF1alpha, thus promoting differentiation or apoptosis of NSCs and progenitors. These dynamic changes in O2 tension can be mimicked to optimize ex vivo production methods for cell replacement therapies. Conversely, disrupted O2 availability may play a critical role in disease states such as stroke or brain tumor progression. Hypoxia during stroke activates precursor proliferation in vivo, while glioblastoma stem cells proliferate maximally in a more hypoxic environment than normal stem cells, which may make them resistant to certain anti-neoplastic therapies. These findings suggest that O2 response is central to the normal architecture and dynamics of NSC regulation and in the etiology and treatment of brain diseases.


Assuntos
Encefalopatias/metabolismo , Neurônios/metabolismo , Oxigênio/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Apoptose , Encefalopatias/patologia , Encefalopatias/cirurgia , Neoplasias Encefálicas/metabolismo , Diferenciação Celular , Hipóxia Celular , Linhagem da Célula , Proliferação de Células , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Neoplásicas/metabolismo , Regeneração Nervosa , Neurônios/patologia , Neurônios/transplante , Transplante de Células-Tronco , Células-Tronco/patologia , Acidente Vascular Cerebral/metabolismo
6.
J Cell Biol ; 161(5): 911-21, 2003 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-12796477

RESUMO

The ability of stem cells to generate distinct fates is critical for the generation of cellular diversity during development. Central nervous system (CNS) stem cells respond to bone morphogenetic protein (BMP) 4 by differentiating into a wide variety of dorsal CNS and neural crest cell types. We show that distinct mechanisms are responsible for the generation of two of these cell types, smooth muscle and glia. Smooth muscle differentiation requires BMP-mediated Smad1/5/8 activation and predominates where local cell density is low. In contrast, glial differentiation predominates at high local densities in response to BMP4 and is specifically blocked by a dominant-negative mutant Stat3. Upon BMP4 treatment, the serine-threonine kinase FKBP12/rapamycin-associated protein (FRAP), mammalian target of rapamycin (mTOR), associates with Stat3 and facilitates STAT activation. Inhibition of FRAP prevents STAT activation and glial differentiation. Thus, glial differentiation by BMP4 occurs by a novel pathway mediated by FRAP and STAT proteins. These results suggest that a single ligand can regulate cell fate by activating distinct cytoplasmic signals.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Proteínas de Ligação a DNA/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool) , Células-Tronco/metabolismo , Transativadores/metabolismo , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Fator Neurotrófico Ciliar/metabolismo , Fator Neurotrófico Ciliar/farmacologia , Proteínas de Ligação a DNA/genética , Feto , Camundongos , Músculo Liso/citologia , Músculo Liso/embriologia , Músculo Liso/metabolismo , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Sistema Nervoso Periférico/citologia , Sistema Nervoso Periférico/embriologia , Sistema Nervoso Periférico/metabolismo , Ratos , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Proteínas Smad , Proteína Smad1 , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Serina-Treonina Quinases TOR , Transativadores/genética
7.
Differentiation ; 76(4): 348-56, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18021260

RESUMO

In eukaryotic cells, covalent modifications to core histones contribute to the establishment and maintenance of cellular phenotype via regulation of gene expression. Histone acetyltransferases (HATs) cooperate with histone deacetylases (HDACs) to establish and maintain specific patterns of histone acetylation. HDAC inhibitors can cause pluripotent stem cells to cease proliferating and enter terminal differentiation pathways in culture. To better define the roles of individual HDACs in stem cell differentiation, we have constructed "dominant-negative" stem cell lines expressing mutant, Flag-tagged HDACs with reduced enzymatic activity. Replacement of a single residue (His-->Ala) in the catalytic center reduced the activity of HDACs 1 and 2 by 80%, and abolished HDAC3 activity; the mutant HDACs were expressed at similar levels and in the same multiprotein complexes as wild-type HDACs. Hexamethylene bisacetamide-induced MEL cell differentiation was potentiated by the individual mutant HDACs, but only to 2%, versus 60% for an HDAC inhibitor, sodium butyrate, suggesting that inhibition of multiple HDACs is required for full potentiation. Cultured E14.5 cortical stem cells differentiate to neurons, astrocytes, and oligodendrocytes upon withdrawal of basic fibroblast growth factor. Transduction of stem cells with mutant HDACs 1, 2, or 3 shifted cell fate choice toward oligodendrocytes. Mutant HDAC2 also increased differentiation to astrocytes, while mutant HDAC1 reduced differentiation to neurons by 50%. These results indicate that HDAC activity inhibits differentiation to oligodendrocytes, and that HDAC2 activity specifically inhibits differentiation to astrocytes, while HDAC1 activity is required for differentiation to neurons.


Assuntos
Diferenciação Celular/fisiologia , Histona Desacetilases/fisiologia , Isoenzimas/fisiologia , Células-Tronco Pluripotentes/citologia , Animais , Domínio Catalítico , Separação Celular , DNA Complementar , Histona Desacetilases/química , Histona Desacetilases/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Mutagênese , Transdução Genética , Células Tumorais Cultivadas
8.
Curr Opin Genet Dev ; 12(4): 478-87, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12100896

RESUMO

A complex orchestration of stem-cell specification, expansion and differentiation is required for the proper development of the nervous system. Although progress has been made on the role of individual genes in each of these processes, there are still unresolved questions about how gene function translates to the dynamic assembly of cells into tissues. Recently, stem-cell biology has emerged as a bridge between the traditional fields of cell biology and developmental genetics. In addition to their potential therapeutic role, stem cells are being exploited as experimental 'logic chips' that integrate information and exhibit self-organizing properties. Recent studies provide new insights on how morphogenic signals coordinate major stem cell decisions to regulate the size, shape and cellular diversity of the nervous system.


Assuntos
Diferenciação Celular/fisiologia , Sistema Nervoso/embriologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Animais , Apoptose/fisiologia , Proteínas de Membrana/fisiologia , Sistema Nervoso/citologia , Receptores Notch , Transcrição Gênica/fisiologia
9.
Stem Cells ; 25(9): 2291-301, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17556599

RESUMO

Despite evidence that oxygen regulates neural precursor fate, the effects of changing oxygen tensions on distinct stages in precursor differentiation are poorly understood. We found that 5% oxygen permitted clonal and long-term expansion of mouse fetal cortical precursors. In contrast, 20% oxygen caused a rapid decrease in hypoxia-inducible factor 1alpha and nucleophosmin, followed by the induction of p53 and apoptosis of cells. This led to a decrease in overall cell number and particularly a loss of astrocytes and oligodendrocytes. Clonal analysis revealed that apoptosis in 20% oxygen was due to a complete loss of CD133(lo)CD24(lo) multipotent precursors, a substantial loss of CD133(hi)CD24(lo) multipotent precursors, and a failure of remaining CD133(hi)CD24(lo) cells to generate glia. In contrast, committed neuronal progenitors were not significantly affected. Switching clones from 5% to 20% oxygen only after mitogen withdrawal led to a decrease in total clone numbers but an even greater decrease in oligodendrocyte-containing clones. During this late exposure to 20% oxygen, bipotent glial (A2B5+) and early (platelet-derived growth factor receptor alpha) oligodendrocyte progenitors appeared and disappeared more quickly, relative to 5% oxygen, and late stage O4+ oligodendrocyte progenitors never appeared. These results indicate that multipotent cells and oligodendrocyte progenitors are more susceptible to apoptosis at 20% oxygen than committed neuronal progenitors. This has important implications for optimizing ex vivo production methods for cell replacement therapies.


Assuntos
Sistema Nervoso Central/citologia , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/embriologia , Oxigênio/farmacologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteína Supressora de Tumor p53/metabolismo
10.
Stem Cell Reports ; 5(6): 933-945, 2015 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-26610635

RESUMO

As a group, we met to discuss the current challenges for creating meaningful patient-specific in vitro models to study brain disorders. Although the convergence of findings between laboratories and patient cohorts provided us confidence and optimism that hiPSC-based platforms will inform future drug discovery efforts, a number of critical technical challenges remain. This opinion piece outlines our collective views on the current state of hiPSC-based disease modeling and discusses what we see to be the critical objectives that must be addressed collectively as a field.


Assuntos
Encefalopatias/patologia , Encéfalo/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Neurogênese , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Encefalopatias/tratamento farmacológico , Encefalopatias/genética , Encefalopatias/fisiopatologia , Descoberta de Drogas/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Mosaicismo , Medicina de Precisão/métodos
11.
Stem Cells Transl Med ; 2(3): 217-22, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23408104

RESUMO

This report synthesizes the discussions during a workshop convened April 24-25, 2012, by the National Institute of Mental Health and the Foundation for the NIH in Bethesda, Maryland, that focused on progress and challenges in the use of patient-derived reprogrammed cells for basic biological discovery, target identification, screening, and drug development for mental illnesses such as schizophrenia, bipolar disorder, and autism spectrum disorders. The workshop revealed that the greatest progress has been made in reprogramming methods and agreed-upon standards for validating the resulting induced pluripotent stem cell lines. However, challenges remain in several areas, including efficiently generating and validating specific neural cell types with respect to regional identity, establishing assays with predictive validity to mental illness pathophysiology, and generating sufficient statistical power and data reproducibility across laboratories. A brainstorming session yielded a number of suggestions, including calls to (a) facilitate the replication of results by standardizing protocols and samples used across laboratories; (b) improve technology by generating cheaper/faster targeting methods, reporters, and assays; and (c) improve resource sharing and collaboration, with an emphasis on rapid sharing of new cell lines, technologies, and best practices, possibly incorporated into a public-private partnership. The meeting provided an important venue for academic, government, and private sector scientists to address potential opportunities for translational and clinical applications of reprogrammed cell research. A number of activities since the workshop have reflected the feedback from meeting participants.


Assuntos
Terapia Genética/métodos , Transtornos Mentais/terapia , Células-Tronco Neurais/transplante , Células-Tronco Pluripotentes/transplante , Animais , Pesquisa Biomédica/normas , Biotecnologia/normas , Reprogramação Celular , Comportamento Cooperativo , Predisposição Genética para Doença , Genótipo , Humanos , Comunicação Interdisciplinar , Cooperação Internacional , Transtornos Mentais/diagnóstico , Transtornos Mentais/genética , Transtornos Mentais/patologia , Neurogênese , Seleção de Pacientes , Fenótipo , Reprodutibilidade dos Testes
12.
Regen Med ; 7(1): 33-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22168495

RESUMO

The nervous system is consistently viewed as a target of high interest for stem cell-based therapeutics. In the USA, the National Institute of Neurological Disorders and Stroke (NINDS) and the National Institute of Mental Health (NIMH) are the two largest funders of neuroscience-related research within the NIH. Research spanning the spectrum of basic, translational and clinical science is conducted both on the NIH campus and through funding of extramural research organizations across the USA, and, to a lesser extent, worldwide. In this brief survey, we will present an overview of stem cell-related activities at the two neuroscience-focused NIH institutes.


Assuntos
National Institute of Mental Health (U.S.) , National Institute of Neurological Disorders and Stroke (USA) , Pesquisa com Células-Tronco , Animais , Humanos , National Institute of Mental Health (U.S.)/economia , National Institute of Neurological Disorders and Stroke (USA)/economia , Neurociências/economia , Neurociências/organização & administração , Pesquisa com Células-Tronco/economia , Estados Unidos
13.
Biochem Pharmacol ; 80(10): 1517-27, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20705058

RESUMO

Glioblastoma multiforme (GBM) are highly proliferative brain tumors characterized by a hypoxic microenvironment which controls GBM stem cell maintenance. Tumor hypoxia promotes also elevated glycolytic rate; thus, limiting glucose metabolism is a potential approach to inhibit tumor growth. Here we investigate the effects mediated by 2-deoxyglucose (2-DG), a glucose analogue, on primary GBM-derived cells maintained under hypoxia. Our results indicate that hypoxia protects GBM cells from the apoptotic effect elicited by 2-DG, which raises succinate dehydrogenase activity thus promoting succinate level decrease. As a consequence hypoxia inducible factor-1α (HIF-1α) degradation occurs and this induces GBM cells to acquire a neuronal committed phenotype. By adding succinate these effects are reverted, as succinate stabilizes HIF-1α and increases GBM stem cell fraction particularly under hypoxia, thus preserving the tumor stem cell niche. 2-DG inhibits anaerobic glycolysis altering GBM cell phenotype by forcing tumor cells into mitochondrial metabolism and by inducing differentiation.


Assuntos
Encéfalo , Desoxiglucose/farmacologia , Glioblastoma , Ácido Succínico/metabolismo , Adulto , Apoptose/efeitos dos fármacos , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Criança , Glioblastoma/metabolismo , Glioblastoma/patologia , Glutationa/metabolismo , Glicólise , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Oxigênio/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Succinato Desidrogenase/metabolismo , Ácido Succínico/farmacologia
14.
PLoS One ; 4(7): e6206, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19587783

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is one of most common and still poorly treated primary brain tumors. In search for new therapeutic approaches, Bone Morphogenetic Proteins (BMPs) induce astroglial commitment in GBM-derived cells in vitro. However, we recently suggested that hypoxia, which is characteristic of the brain niche where GBM reside, strongly counter-acts BMP effects. It seems apparent that a more complete understanding of the biology of GBM cells is needed, in particular considering the role played by hypoxia as a signaling pathways regulator. HIF-1alpha is controlled at the transcriptional and translational level by mTOR and, alike BMP, also mTOR pathway modulates glial differentiation in central nervous system (CNS) stem cells. METHODOLOGY/PRINCIPAL FINDINGS: Here, we investigate the role of mTOR signaling in the regulation of HIF-1alpha stability in primary GBM-derived cells maintained under hypoxia (2% oxygen). We found that GBM cells, when acutely exposed to high oxygen tension, undergo Akt/mTOR pathway activation and that BMP2 acts in an analogous way. Importantly, repression of Akt/mTOR signaling is maintained by HIF-1alpha through REDD1 upregulation. On the other hand, BMP2 counter-acts HIF-1alpha stability by modulating intracellular succinate and by controlling proline hydroxylase 2 (PHD2) protein through inhibition of FKBP38, a PHD2 protein regulator. CONCLUSIONS/SIGNIFICANCE: In this study we elucidate the molecular mechanisms by which two pro-differentiating stimuli, BMP2 and acute high oxygen exposure, control HIF-1alpha stability. We previously reported that both these stimuli, by inducing astroglial differentiation, affect GBM cells growth. We also found differences in high oxygen and BMP2 sensitivity between GBM cells and normal cells that should be further investigated to better define tumor cell biology.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Oxigênio/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Cobalto/farmacologia , Inativação Gênica , Glioblastoma/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR , Proteínas de Ligação a Tacrolimo/metabolismo
15.
Cell Stem Cell ; 3(6): 670-80, 2008 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19041783

RESUMO

To realize the promise of stem cell biology, it is important to identify the precise time in the history of the cell when developmental potential is restricted. To achieve this goal, we developed a real-time imaging system that captures the transitions in fate, generating neurons, astrocytes, and oligodendrocytes from single CNS stem cells in vitro. In the presence of bFGF, tripotent cells normally produce specified progenitors through a bipotent intermediate cell type. Surprisingly, the tripotent state is reset at each passage. The cytokine CNTF is thought to instruct multipotent cells to an astrocytic fate. We demonstrate that CNTF both directs astrogliogenesis from tripotent cells, bypassing two of the three normal bipotent intermediates, and later promotes the expansion of specified astrocytic progenitors. These results show how discrete cell types emerge from a multipotent cell and provide a strong basis for future studies to determine the molecular basis of fate specification.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Sistema Nervoso Central/embriologia , Células-Tronco Multipotentes/metabolismo , Neurogênese/fisiologia , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Fator Neurotrófico Ciliar/metabolismo , Fator Neurotrófico Ciliar/farmacologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Genes Reporter/genética , Proteína Glial Fibrilar Ácida/genética , Citometria por Imagem/métodos , Microscopia de Vídeo/métodos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Neurônios/citologia , Neurônios/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Ratos , Ratos Sprague-Dawley , Técnicas de Cultura de Tecidos/métodos , Transfecção/métodos
16.
Stem Cells ; 25(1): 63-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16973830

RESUMO

Bone morphogenetic proteins (BMPs) are a class of morphogens that are critical regulators of the central nervous system (CNS), peripheral nervous system, and craniofacial development. Modulation of BMP signaling also appears to be an important component of the postnatal stem cell niche. However, describing a comprehensive model of BMP actions is complicated by their paradoxical effects in precursor cells, which include dorsal specification, promoting proliferation or mitotic arrest, cell survival or death, and neuronal or glial fate. In addition, in postmitotic neurons BMPs can promote dendritic growth, act as axonal chemorepellants, and stabilize synapses. Although many of these responses depend on interactions with other incoming signals, some reflect the recruitment of distinct BMP signal transduction pathways. In this review, we classify the diverse effects of BMPs on neural cells, focus on the known mechanisms that specify distinct responses, and discuss the remaining challenges in identifying the cellular basis of BMP pleiotropism. Addressing these issues may have importance for stem cell mobilization, differentiation, and cell integration/survival in reparative therapies.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Divisão Celular , Desenvolvimento Embrionário , Humanos , Modelos Biológicos , Morfogênese
17.
Stem Cells ; 25(6): 1560-70, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17332513

RESUMO

Although flow cytometry is useful for studying neural lineage relationships, the method of dissociation can potentially bias cell analysis. We compared dissociation methods on viability and antigen recognition of mouse central nervous system (CNS) tissue and human CNS tumor tissue. Although nonenzymatic dissociation yielded poor viability, papain, purified trypsin replacement (TrypLE), and two purified collagenase/neutral protease cocktails (Liberase-1 or Accutase) each efficiently dissociated fetal tissue and postnatal tissue. Mouse cells dissociated with Liberase-1 were titrated with antibodies identifying distinct CNS precursor subtypes, including CD133, CD15, CD24, A2B5, and PSA-NCAM. Of the enzymes tested, papain most aggressively reduced antigenicity for mouse and human CD24. On human CNS tumor cells, CD133 expression remained highest after Liberase-1 and was lowest after papain or Accutase treatment; Liberase-1 digestion allowed magnetic sorting for CD133 without the need for an antigen re-expression recovery period. We conclude that Liberase-1 and TrypLE provide the best balance of dissociation efficiency, viability, and antigen retention. One implication of this comparison was confirmed by dissociating E13.5 mouse cortical cells and performing prospective isolation and clonal analysis on the basis of CD133/CD24 or CD15/CD24 expression. Highest fetal expression of CD133 or CD15 occurred in a CD24(hi) population that was enriched in neuronal progenitors. Multipotent cells expressed CD133 and CD15 at lower levels than did these neuronal progenitors. We conclude that CD133 and CD15 can be used similarly as selectable markers, but CD24 coexpression helps to distinguish fetal mouse multipotent stem cells from neuronal progenitors and postmitotic neurons. This particular discrimination is not possible after papain treatment. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Antígenos CD/metabolismo , Antígeno CD24/metabolismo , Separação Celular/métodos , Sistema Nervoso Central/citologia , Citometria de Fluxo , Glicoproteínas/metabolismo , Antígenos CD15/metabolismo , Tecido Nervoso/citologia , Peptídeos/metabolismo , Antígeno AC133 , Animais , Animais Recém-Nascidos , Antígenos de Superfície/análise , Neoplasias Encefálicas/patologia , Carcinoma/patologia , Células Cultivadas , Sistema Nervoso Central/embriologia , Criança , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Tecido Nervoso/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
18.
Mol Cell Neurosci ; 35(3): 424-35, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17498968

RESUMO

Human neural precursor proliferation and potency is limited by senescence and loss of oligodendrocyte potential. We found that in vitro expansion of human postnatal brain CD133(+) nestin(+) precursors is enhanced at 5% oxygen, while raising oxygen tension to 20% depletes precursors and promotes astrocyte differentiation even in the presence of mitogens. Higher cell densities yielded more astrocytes regardless of oxygen tension. This was reversed by noggin at 5%, but not 20%, oxygen due to a novel repressive effect of low oxygen on bone morphogenetic protein (BMP) signaling. When induced to differentiate by mitogen withdrawal, 5% oxygen-expanded precursors generated 17-fold more oligodendrocytes than cells expanded in 20% oxygen. When precursors were expanded at 5% oxygen and then differentiated at 20% oxygen, oligodendrocyte maturation was further enhanced 2.5-fold. These results indicate that dynamic control of oxygen tension regulates different steps in fate and maturation and may be crucial for treating neurodegenerative diseases.


Assuntos
Astrócitos/efeitos dos fármacos , Sistema Nervoso Central/citologia , Oligodendroglia/efeitos dos fármacos , Oxigênio/farmacologia , Células-Tronco/efeitos dos fármacos , Análise de Variância , Antígenos CD/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/crescimento & desenvolvimento , Relação Dose-Resposta a Droga , Feto , Citometria de Fluxo/métodos , Humanos , Recém-Nascido , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco/fisiologia , Fatores de Tempo
19.
J Cell Sci ; 118(Pt 24): 5849-60, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16339968

RESUMO

CNS stem cells are best characterized by their ability to self-renew and to generate multiple differentiated derivatives, but the effect of mitogenic signals, such as fibroblast growth factor 2 (FGF2), on the positional identity of these cells is not well understood. Here, we report that bone morphogenetic protein 2 (BMP2) induces telencephalic CNS stem cells to fates characteristic of neural crest and choroid plexus mesenchyme, a cell type of undetermined lineage in rodents. This induction occurs both in dissociated cell culture and cortical explants of embryonic day 14.5 (E14.5) embryos, but only when cells have been exposed to FGF2. Neither EGF nor IGF1 can substitute for FGF2. An early step in this response is activation of beta-catenin, a mediator of Wnt activity. The CNS stem cells first undergo an epithelial-to-mesenchymal transition and subsequently differentiate to smooth-muscle and non-CNS glia cells. Similar responses are seen with stem cells from E14.5 cortex, E18.5 cortex and adult subventricular zone, but with a progressive shift toward gliogenesis that is characteristic of normal development. These data indicate that FGF2 confers competence for dorsalization independently of its mitogenic action. This rapid and efficient induction of dorsal fates may allow identification of positional identity effectors that are co-regulated by FGF2 and BMP2.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem da Célula/fisiologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Crista Neural/embriologia , Células-Tronco/metabolismo , Telencéfalo/embriologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteína Morfogenética Óssea 2 , Diferenciação Celular/fisiologia , Plexo Corióideo/citologia , Plexo Corióideo/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Crista Neural/citologia , Ratos , Telencéfalo/citologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA